MTA2 (Metastasis-associated protein 2), also known as MTA1L1 or PID, is a 668 amino acid protein with a molecular weight of approximately 75 kDa. It plays a crucial role in gene expression regulation as a component of the nucleosome remodeling and histone deacetylation (NURD/NuRD) complex . The significance of MTA2 as a research target stems from its involvement in ATP-dependent chromatin remodeling and transcriptional repression, influencing cellular processes including differentiation and proliferation . MTA2's ability to modulate chromatin structure and gene expression makes it important in both normal cellular function and disease states, particularly in cancer progression .
| Application | Recommended Antibody Type | Optimal Dilution Range | Species Reactivity |
|---|---|---|---|
| Western Blot (WB) | Rabbit polyclonal (17554-1-AP) | 1:2000-1:10000 | Human, Mouse, Rat |
| Mouse monoclonal (66195-1-Ig) | 1:5000-1:50000 | Human, Mouse, Rat | |
| Immunohistochemistry (IHC) | Rabbit polyclonal (17554-1-AP) | 1:500-1:2000 | Human, Mouse, Rat |
| Mouse monoclonal (66195-1-Ig) | 1:500-1:2000 | Human, Mouse, Rat | |
| Immunofluorescence (IF) | Mouse monoclonal (66195-1-Ig) | 1:1000-1:2000 | Human, Mouse, Rat |
| Immunoprecipitation (IP) | Rabbit polyclonal (17554-1-AP) | 0.5-4.0 μg for 1-3 mg lysate | Human, Mouse, Rat |
| ChIP | Various | Application-specific | Human, Mouse |
For optimal results, consider the following methodological guidelines:
For WB applications requiring high sensitivity, mouse monoclonal antibodies (66195-1-Ig) provide superior results with less background at higher dilutions
For IHC applications, antigen retrieval with TE buffer pH 9.0 is recommended, though citrate buffer pH 6.0 may be used as an alternative
For multiplex applications, select antibodies from different host species to prevent cross-reactivity
Optimizing antibody concentration for Western blotting requires a systematic approach:
Start with a medium dilution range (e.g., 1:5000) from the manufacturer's recommendation
Perform a dilution series experiment: For polyclonal antibodies (17554-1-AP), test dilutions between 1:2000-1:10000; for monoclonal antibodies (66195-1-Ig), test between 1:5000-1:50000
Evaluate signal-to-noise ratio at each concentration
Consider the cell/tissue type being analyzed - different samples may require different optimal concentrations
For weak signals, particularly with endogenous MTA2, lower dilutions (higher antibody concentration) may be necessary, but monitor background
Implement proper blocking (5% non-fat milk in TBS is effective)
Secondary antibody optimization is also crucial - for rabbit primaries, goat anti-rabbit IgG-HRP at 1:5000 provides good results
Methodological observation: Mouse monoclonal antibodies typically allow for higher dilutions while maintaining specificity, which can be cost-effective for long-term research projects .
Based on validated research data, the following positive controls are recommended for MTA2 antibody validation:
For proper validation methodology:
Include at least one well-characterized positive control in each experiment
Load approximately 50 μg of protein lysate per lane for optimal detection
Compare observed molecular weight (70-75 kDa) with predicted weight (75 kDa)
For definitive validation, include MTA2 knockdown/knockout samples as negative controls
For tissue samples, confirm appropriate antigen retrieval method before concluding negative results
MTA2 functions as a key component of the nucleosome remodeling and histone deacetylation (NURD/NuRD) complex. To study these interactions:
Co-Immunoprecipitation (Co-IP):
Chromatin Immunoprecipitation (ChIP):
Proximity Ligation Assay (PLA):
Combine MTA2 antibody with antibodies against other complex components
This technique visualizes protein-protein interactions in situ with high specificity
Sequential ChIP (Re-ChIP):
First ChIP with MTA2 antibody followed by second ChIP with antibodies against other NURD/NuRD components
Identifies genomic regions where multiple complex members co-localize
Methodological consideration: When studying protein-protein interactions, antibody orientation is crucial - the epitope recognized by the antibody should not interfere with the interaction interface of interest .
MTA2 has been shown to deacetylate p53, affecting p53-dependent transcriptional activation and modulating cell growth arrest and apoptosis. Here are methodological approaches to investigate this relationship:
Acetylation Status Analysis:
Immunoprecipitate p53 using specific antibodies
Probe with acetylation-specific antibodies (e.g., anti-acetyl-p53)
Compare acetylation levels in cells with normal vs. altered MTA2 expression
MTA2 Modulation:
Functional Assays:
Cell cycle analysis: Flow cytometry to determine if MTA2 manipulation alters p53-dependent cell cycle arrest
Apoptosis assays: Annexin V staining, TUNEL assay to measure p53-dependent apoptosis
Reporter assays: Use p53-responsive luciferase reporters to measure transcriptional activity
In vivo Models:
Clinical Correlation:
Analyze MTA2 expression in patient samples using validated antibodies
Correlate with p53 mutation status, acetylation levels, and clinical outcomes
Technical note: When performing IHC on clinical samples, antigen retrieval with TE buffer (pH 9.0) provides optimal results, though citrate buffer (pH 6.0) may be used as an alternative .
Advanced multiplexed imaging techniques allow for simultaneous visualization of multiple proteins in their spatial context. For MTA2 studies:
Multiplex Immunofluorescence:
Use mouse monoclonal MTA2 antibody (66195-1-Ig) at 1:1000-1:2000 dilution
Combine with antibodies against other proteins of interest from different host species
Select fluorophore-conjugated secondary antibodies with minimal spectral overlap
For nuclear proteins like MTA2, confocal microscopy provides superior resolution
Cyclic Immunofluorescence (CycIF):
Apply MTA2 antibody in one cycle, image, then strip/quench fluorescence
Repeat with different antibodies in subsequent cycles
Enables visualization of >30 proteins on the same sample
Critical for studying MTA2 in relation to multiple signaling pathways
Mass Cytometry Imaging (IMC):
Label MTA2 antibodies with rare earth metal isotopes
Allows for simultaneous detection of >40 proteins with minimal signal overlap
Particularly useful for tumor microenvironment studies
Proximity Ligation Assay (PLA) in Tissue Sections:
Combine MTA2 antibody with antibodies against potential interacting partners
Provides spatial information about protein-protein interactions in situ
Methodological considerations:
For multiplexed approaches, validate antibodies individually before combining
Include appropriate controls for each antibody in the panel
MTA2 antibodies from mouse hosts (66195-1-Ig) work well in multiplexed systems with rabbit antibodies against other targets
Counterstain nuclei with DAPI to facilitate identification of MTA2-positive nuclei
Inconsistent staining patterns in IHC can arise from multiple factors. Here's a systematic troubleshooting approach:
Antigen Retrieval Optimization:
Fixation Variables:
Overfixation can mask epitopes - standardize fixation times
For FFPE tissues, section thickness should be consistent (4-5 μm)
Consider testing different fixatives if possible
Antibody Dilution Series:
Detection System Evaluation:
Compare different visualization methods:
DAB-based detection systems (brown signal)
AP-based systems (red signal) may give cleaner results in some tissues
For fluorescent detection, test different fluorophores and mounting media
Background Reduction:
Methodological note: When comparing staining patterns between different antibodies, process slides in parallel under identical conditions to minimize technical variability .
When encountering weak or absent MTA2 signals in Western blotting, consider these methodological interventions:
Sample Preparation Optimization:
Transfer Efficiency Improvement:
Antibody Selection and Handling:
Signal Enhancement Techniques:
Use high-sensitivity ECL substrates
Increase exposure time incrementally
For weak signals, consider HRP-conjugated secondary antibodies with higher activity
Membrane Handling:
Positive Control Inclusion:
Experimental observation: MTA2 signal can sometimes appear as a doublet around 70-75 kDa, representing different post-translational modification states or isoforms .
The MTA family includes MTA1, MTA2, and MTA3, which share significant sequence homology. To ensure specificity for MTA2:
Antibody Selection Criteria:
Western Blot Differentiation:
qPCR Validation:
Complement protein-level data with mRNA analysis
Design primers specific to unique regions of MTA2
Use this as orthogonal validation of antibody specificity
Knockout/Knockdown Controls:
Epitope Mapping:
Immunoprecipitation-Mass Spectrometry:
For definitive identification, perform IP with MTA2 antibody
Analyze immunoprecipitated proteins by mass spectrometry
This can confirm the presence of MTA2 and detect potential cross-reactivity
Technical consideration: For applications requiring absolute specificity, monoclonal antibodies generally offer better discrimination between MTA family members than polyclonal antibodies .
When analyzing MTA2 expression patterns across tissues, consider these methodological approaches and interpretive frameworks:
Baseline Expression Profile:
Normalization Strategies:
For Western blot analysis:
Normalize to total protein (Ponceau S or REVERT staining)
Use multiple housekeeping proteins as loading controls
Consider nuclear-specific loading controls for nuclear proteins like MTA2
For IHC/IF quantification:
Express as percentage of positive cells
Use H-score or Allred scoring systems for semi-quantitative assessment
Employ digital image analysis for objective quantification
Biological Interpretation Framework:
Higher expression in specific tissues may indicate:
Tissue-specific regulatory functions
Association with proliferative capacity of the tissue
Involvement in tissue-specific developmental programs
Changes during disease states may reflect:
Altered chromatin regulation
Modified transcriptional programs
Response to cellular stress
Technical Variables to Consider:
Different fixation methods across tissue types can affect epitope availability
Tissue-specific protein extraction efficiency may vary
Endogenous peroxidase activity varies by tissue (relevant for IHC)
Validation Through Orthogonal Methods:
Confirm protein-level observations with mRNA analysis
Compare antibody-based detection with reporter systems
Cross-validate with public databases (Human Protein Atlas, TCGA)
Methodological note: For accurate comparison across tissues, process all samples simultaneously using identical protocols, and include internal reference tissues in each experiment .
MTA2 functions primarily in the nucleus as part of chromatin-modifying complexes, but proper analysis of its subcellular localization requires careful consideration:
Fixation and Permeabilization Optimization:
For nuclear proteins like MTA2:
Paraformaldehyde (4%) fixation preserves nuclear architecture
Methanol fixation can improve nuclear antigen accessibility
Permeabilization is crucial - test Triton X-100 (0.1-0.5%) or saponin (0.1%)
Counterstaining Strategy:
Quantitative Analysis Approaches:
Measure nuclear:cytoplasmic ratio of MTA2 signal
Assess co-localization with other nuclear proteins (Pearson's correlation)
Evaluate subnuclear distribution patterns (diffuse vs. punctate)
Consider 3D reconstruction from confocal z-stacks for complete analysis
Dynamic Localization Considerations:
MTA2 localization may change with:
Cell cycle stage
Differentiation status
Stress conditions
Treatment with chromatin-modifying agents
Time-course experiments may reveal regulatory mechanisms
Technical Controls for Localization Studies:
Include proteins with known localization patterns
Test multiple antibodies targeting different epitopes
Verify with fluorescent protein-tagged MTA2 constructs
Use fractionation followed by Western blot as orthogonal validation
Methodological insight: For immunofluorescence applications, confocal microscopy with deconvolution provides superior resolution of nuclear proteins like MTA2, particularly when examining co-localization with other nuclear factors .
Discriminating between technical artifacts and genuine biological variability requires systematic controls and analysis:
Comprehensive Control System:
Positive technical controls:
Negative technical controls:
Primary antibody omission
Isotype control antibodies
Blocking peptide competition
Biological validation controls:
Batch Effect Minimization:
Process all comparative samples in parallel
Maintain identical conditions for all technical steps
Include standard reference samples across batches
Consider multiplexed approaches to reduce inter-slide variability
Pattern Recognition Framework:
Technical artifacts often present as:
Edge effects or drying artifacts
Uniform background across all cell types
Signal in unexpected subcellular locations
Biological patterns typically show:
Cell type-specific expression
Correlation with differentiation/proliferation status
Appropriate subcellular localization (nuclear for MTA2)
Correlation with functionally related proteins
Orthogonal Validation Strategies:
Confirm key findings with alternative antibodies
Validate with non-antibody methods (RNA-seq, reporter assays)
Compare with public database information
Use multiple detection methods (IF vs. IHC vs. WB)
Quantitative Analysis Approaches:
Establish objective scoring criteria before analysis
Use digital image analysis when possible
Apply statistical methods appropriate for the data distribution
Consider blinded assessment by multiple observers
Technical insight: When troubleshooting inconsistent results, systematically vary one parameter at a time (fixation, antigen retrieval, antibody concentration) while keeping others constant to identify the source of variability .
As single-cell technologies revolutionize biological research, MTA2 antibodies can be integrated into these emerging platforms:
Single-Cell Western Blotting:
Mass Cytometry (CyTOF):
MTA2 antibodies can be conjugated to rare earth metals
Allows simultaneous detection of MTA2 with >40 other proteins
Critical for understanding MTA2 in complex cellular hierarchies
No fluorescence overlap issues compared to flow cytometry
Single-Cell Proteomics with Antibody-Based Capture:
MTA2 antibodies immobilized on microwell arrays
Capture proteins from lysed single cells
Detection via complementary antibodies or mass spectrometry
Reveals MTA2 protein interaction networks at single-cell resolution
In situ Protein Analysis:
Techniques like CODEX use DNA-barcoded antibodies for spatial proteomics
MTA2 antibodies can be incorporated into multiplexed panels
Preserves tissue architecture while achieving single-cell resolution
Particularly valuable for understanding MTA2 in tumor microenvironments
Proximity Ligation Assay at Single-Cell Level:
Combine MTA2 antibodies with antibodies against interaction partners
Detect specific protein-protein interactions in individual cells
Quantify interaction frequencies across heterogeneous populations
Methodological consideration: For single-cell applications, antibody validation is even more critical - perform extensive specificity testing, including isotype controls and blocking peptide competition assays before application in these sensitive techniques .
MTA2 function is regulated by various post-translational modifications (PTMs). Studying these modifications requires specialized approaches:
Modification-Specific Antibody Selection:
For known MTA2 PTMs (phosphorylation, acetylation, SUMOylation)
Requires validation of modification-specific antibodies
Consider generating custom antibodies if commercial options unavailable
Enrichment Strategies:
Mass Spectrometry Workflow:
Immunoprecipitate MTA2 with validated antibodies
Perform tryptic digestion
Analyze by LC-MS/MS with PTM-specific detection methods
Consider enrichment steps for low-abundance modifications
Functional Correlation Analysis:
Compare PTM status across different:
Cell types
Cellular conditions (stress, differentiation)
Disease states
Correlate with MTA2 activity measurements
Test effects of PTM-mimicking or PTM-blocking mutations
Dynamic PTM Regulation Studies:
Time-course experiments following stimuli
Inhibitor studies targeting specific modifying enzymes
Consider proximity labeling approaches to identify regulatory enzymes
Quantitative PTM Analysis:
Use SILAC or TMT labeling for quantitative mass spectrometry
Develop standard curves with synthetic phosphopeptides/acetylpeptides
Compare stoichiometry across conditions
Methodological note: When analyzing PTMs, sample handling is critical - include phosphatase inhibitors, deacetylase inhibitors, and deSUMOylase inhibitors in lysis buffers to preserve modification status .
Investigating MTA2's role in chromatin dynamics and 3D genome architecture requires integrating antibody-based approaches with advanced genomic technologies:
Chromatin Immunoprecipitation Methodologies:
3D Genome Architecture Analysis:
ChIA-PET using MTA2 antibodies to identify long-range chromatin interactions
HiChIP to combine Hi-C with MTA2 ChIP for interaction mapping
Integrate with ATAC-seq to correlate MTA2 binding with chromatin accessibility
4C-seq focused on MTA2-bound regions as viewpoints
Multi-Omics Integration:
Combine MTA2 ChIP-seq with:
RNA-seq to correlate binding with gene expression
Histone modification ChIP-seq to understand chromatin state
DNA methylation analysis for epigenetic context
Requires computational integration of multiple datasets
Live-Cell Chromatin Dynamics:
Adaptation of MTA2 antibodies for live-cell imaging:
Nanobody development based on validated antibodies
Cell-permeable antibody fragments
Alternative: CRISPR-based tagging of endogenous MTA2
Super-Resolution Microscopy Applications:
Technical consideration: For ChIP applications with MTA2 antibodies, crosslinking conditions are critical - standard 1% formaldehyde may be insufficient for proteins in large complexes like NURD/NuRD; consider testing dual crosslinkers (formaldehyde plus disuccinimidyl glutarate) for improved efficiency .